28
1 Universality of cell differentiation trajectories revealed by a reconstruction of transcriptional uncertainty landscapes from single-cell transcriptomic data Nan Papili Gao 1 , Olivier Gandrillon 2,3 , András Páldi 4 , Ulysse Herbach 5 , Rudiyanto Gunawan 6,* 1 Institute for Chemical and Bioengineering, ETH Zurich, 8093 Zurich, Switzerland 2 Univ Lyon, ENS de Lyon, Univ Claude Bernard, CNRS UMR 5239, INSERM U1210, Laboratory of Biology and Modelling of the Cell, F69007 Lyon, France 3 Inria Team Dracula, Inria Center Grenoble Rhône-Alpes, France 4 Ecole Pratique des Hautes Etudes, PSL Research University, St-Antoine Research Center, Inserm U938, 34 rue Crozatier, 75012, Paris, France 5 Université de Lorraine, CNRS, Inria, IECL, F-54000 Nancy, France 6 Department of Chemical and Biological Engineering, University at Buffalo, Buffalo, NY 14260, USA *Corresponding author (email: [email protected]) ABSTRACT We employed our previously-described single-cell gene expression analysis CALISTA (Clustering And Lineage Inference in Single-Cell Transcriptional Analysis) to evaluate transcriptional uncertainty at the single-cell level using a stochastic mechanistic model of gene expression. We reconstructed a transcriptional uncertainty landscape during cell differentiation by visualizing single-cell transcriptional uncertainty surface over a two dimensional representation of the single-cell gene expression data. The reconstruction of transcriptional uncertainty landscapes for ten publicly available single-cell gene expression datasets from cell differentiation processes with linear, single or multi-branching cell lineage, reveals universal features in the cell differentiation trajectory that include: (i) a peak in single-cell uncertainty during transition states, and in systems with bifurcating differentiation trajectories, each branching point represents a state of high transcriptional uncertainty; (ii) a positive correlation of transcriptional uncertainty with transcriptional burst size and frequency; (iii) an increase in RNA velocity preceeding the increase in the cell transcriptional uncertainty. Finally, we provided biological interpretations of the universal rise-then-fall profile of the transcriptional uncertainty landscape, including a link with the Waddington’s epigenetic landscape, that is generalizable to every cell differentiation system. Keywords: single cell, cell differentiation, epigenetic landscape, gene expression, heterogeneity . CC-BY-NC-ND 4.0 International license (which was not certified by peer review) is the author/funder. It is made available under a The copyright holder for this preprint this version posted April 25, 2020. . https://doi.org/10.1101/2020.04.23.056069 doi: bioRxiv preprint

Universality of cell differentiation trajectories revealed by a … · 1 Universality of cell differentiation trajectories revealed by a reconstruction of transcriptional uncertainty

  • Upload
    others

  • View
    3

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Universality of cell differentiation trajectories revealed by a … · 1 Universality of cell differentiation trajectories revealed by a reconstruction of transcriptional uncertainty

1

Universality of cell differentiation trajectories revealed by

a reconstruction of transcriptional uncertainty landscapes

from single-cell transcriptomic data

Nan Papili Gao1, Olivier Gandrillon 2,3, András Páldi 4, Ulysse Herbach 5, Rudiyanto Gunawan 6,*

1 Institute for Chemical and Bioengineering, ETH Zurich, 8093 Zurich, Switzerland

2 Univ Lyon, ENS de Lyon, Univ Claude Bernard, CNRS UMR 5239, INSERM U1210, Laboratory of Biology

and Modelling of the Cell, F69007 Lyon, France

3 Inria Team Dracula, Inria Center Grenoble Rhône-Alpes, France

4 Ecole Pratique des Hautes Etudes, PSL Research University, St-Antoine Research Center, Inserm U938, 34 rue

Crozatier, 75012, Paris, France

5 Université de Lorraine, CNRS, Inria, IECL, F-54000 Nancy, France

6 Department of Chemical and Biological Engineering, University at Buffalo, Buffalo, NY 14260, USA

*Corresponding author (email: [email protected])

ABSTRACT

We employed our previously-described single-cell gene expression analysis CALISTA (Clustering And Lineage

Inference in Single-Cell Transcriptional Analysis) to evaluate transcriptional uncertainty at the single-cell level

using a stochastic mechanistic model of gene expression. We reconstructed a transcriptional uncertainty landscape

during cell differentiation by visualizing single-cell transcriptional uncertainty surface over a two dimensional

representation of the single-cell gene expression data. The reconstruction of transcriptional uncertainty landscapes

for ten publicly available single-cell gene expression datasets from cell differentiation processes with linear, single

or multi-branching cell lineage, reveals universal features in the cell differentiation trajectory that include: (i) a

peak in single-cell uncertainty during transition states, and in systems with bifurcating differentiation trajectories,

each branching point represents a state of high transcriptional uncertainty; (ii) a positive correlation of

transcriptional uncertainty with transcriptional burst size and frequency; (iii) an increase in RNA velocity

preceeding the increase in the cell transcriptional uncertainty. Finally, we provided biological interpretations of

the universal rise-then-fall profile of the transcriptional uncertainty landscape, including a link with the

Waddington’s epigenetic landscape, that is generalizable to every cell differentiation system.

Keywords: single cell, cell differentiation, epigenetic landscape, gene expression, heterogeneity

.CC-BY-NC-ND 4.0 International license(which was not certified by peer review) is the author/funder. It is made available under aThe copyright holder for this preprintthis version posted April 25, 2020. . https://doi.org/10.1101/2020.04.23.056069doi: bioRxiv preprint

Page 2: Universality of cell differentiation trajectories revealed by a … · 1 Universality of cell differentiation trajectories revealed by a reconstruction of transcriptional uncertainty

2

MAIN

Cell differentiation is the process through which unspecialized stem cells become more specialized. Because

of its importance in development, cellular repair, and organismal homeostasis, the molecular mechanisms of cell

differentiation has been the subject of intense scrutiny. Since roughly 50 years ago – along with the promulgation

of the central dogma of molecular biology by Francis Crick and the characterization of the lactose operon by

François Jacob and Jacques Monod – the existence of a genetic program has become a prevailing explanation for

the cell differentiation process. Although the details were originally not defined, at least not formally, such a

genetic program purports a constellation of master genes (i.e., transcription factors) that orchestrate the

transcription of downstream target genes in a precise spatiotemporal fashion, resulting in long-lasting alterations

in the gene expression patterns 1–3. A notable experimental evidence substantiating this view is the overexpression

of myoD inducing a myogenic phenotype in seemingly naive cells 4. Over the past few decades, the repertoire of

such master genes across numerous stem cell systems, such as Nanog, Oct4, Sox2, BATF and MyoD, begin to

coalesce 5–7.

Recent advances in single-cell technologies has revealed new aspects of the cell differentiation that are

incompatible with the idea of ordered and programmed (i.e., deterministic) gene expression. More specifically,

single-cell data paint a stochastic differentiation process that increases cell-to-cell variability of gene expression.

Such an observation has been made for a wide variety of cell differentiation systems, including chicken erythroid

progenitors 8, erythroid myeloid lymphoid (EML) cells 9, mouse embryonic stem cells (mESCs) 10,11, and human

CD34+ cells 12. Interestingly, a similar increase of gene expression variation was also observed during the de-

differentiation of somatic cells into iPSCs 13. Stochastic gene expression also appears to have functional role

beyond cell differentiation systems. For example, an increase in cell-to-cell variability of gene expression has

been reported during a forced adaptation of budding yeast cells to unforeseen challenges 14.

Based on the observations of single-cell data above, a different view of cell differentiation begins to

materialize. Instead of stem cells following an identical genetic program, the cell differentiation is akin to a

dynamic exploratory process. More specifically, in this view, the cell differentiation is thought to proceed as

follows 14–17:

I) extrinsic and/intrinsic internal stimuli, such as a medium change or the addition of new molecules in the external

medium, trigger a cellular response that destabilizes the initial high potency cell state;

.CC-BY-NC-ND 4.0 International license(which was not certified by peer review) is the author/funder. It is made available under aThe copyright holder for this preprintthis version posted April 25, 2020. . https://doi.org/10.1101/2020.04.23.056069doi: bioRxiv preprint

Page 3: Universality of cell differentiation trajectories revealed by a … · 1 Universality of cell differentiation trajectories revealed by a reconstruction of transcriptional uncertainty

3

II) each cell alters its internal cell state and engages an exploratory dynamics through a combination of the inherent

stochastic dynamics of gene transcription and the emergence of new stable cell state(s). At the cell population

level, we observe a rise in the cell-to-cell variability of gene expression;

III) a physiological selection / commitment to one stable lineage among possibly multiple lineages that arise from

the degeneracy of the system;

IV) finally, a reduction in the exploratory dynamics commences along with the establishment of stable cell state(s)

corresponding to differentiated cell type(s).

The above view is compatible with the idea that cell phenotype transition results from the dynamics of an

underlying stochastic molecular network 18,19. In 1957, Conrad Waddington proposed the presently well-known

epigenetic landscape that likens the cell differentiation process to a ball rolling on a downward sloping surface,

starting from a state of high cell potency and ending at one of possibly several states of low cell potency. The

landscape itself is shaped by the action of the genes and gene network – depicted in the less-frequently-shown

part B of Waddington’s original figure as a network of ropes that are tied to the surface, creating valleys and hills.

Although the epigenetic landscape was originally proposed only as a metaphor of how gene regulation governs

the cell differentiation process, this landscape has been formalized within the framework of dynamical systems

theory 20. The valleys in the Waddington’s epigenetic landscape are equated to stable states of a dynamical system,

called attractors, while the hills are often interpreted as energetic barriers.

The Waddington’s metaphor has further been re-examined in a series of works, where the behaviour of the

underlying dynamical gene regulatory network (GRN) drives cell fate determination. Some of these studies

employed stochastic simulations of simple gene networks with bi-/tri-stable states to describe the role of non-

genetic cell heterogeneity in cell differentiation processes 21–23. Here, to quantitatively reconstruct the epigenetic

landscape, an underlying GRN structure must be assumed a priori. In reality, the GRN driving cell fate

determination is complex and proved to be challenging to infer from data 6,24–28. Even when the complete

regulatory system is known, the high-dimensionality of the parameter space makes the landscape generation

computationally prohibitive 21, especially in the absence of an analytical solution, which are slowly emerging 29.

Thus, the above approaches provide more of a conceptual understanding than a mechanistic or molecular

explanation.

A number of recent studies provided a graphical representation of the differentiation process based on single-

cell transcriptomic data that conforms with the Waddington’s epigenetic landscape 30–34. More specifically, these

studies reconstructed the epigenetic landscape from single-cell gene expression data using probabilistic and quasi-

.CC-BY-NC-ND 4.0 International license(which was not certified by peer review) is the author/funder. It is made available under aThe copyright holder for this preprintthis version posted April 25, 2020. . https://doi.org/10.1101/2020.04.23.056069doi: bioRxiv preprint

Page 4: Universality of cell differentiation trajectories revealed by a … · 1 Universality of cell differentiation trajectories revealed by a reconstruction of transcriptional uncertainty

4

potential methods, for example by applying Hopfield neural networks 30,31, a cell-density based strategy 33,

network entropy measurements 34 or more recently Large Deviation Theory 29. However, with the exception of

Fard et al. 31 and Lv et al., 2014 29, the aforementioned studies produced monotonic descent passages during cell

differentiation, mimicking closely the Waddington’s epigenetic landscape metaphor (see for example 22,34). Also,

none of the above studies consider directly the cellular mechanism that generates stochastic gene transcriptional

bursts.

In the present work, we aimed to shed light on the gene transcriptional mechanism behind the rise-then-fall

trajectory of cell-to-cell variability in gene expression observed during the cellular differentiation process. To this

end, we analyzed a collection (8) of published single-cell transcriptomic datasets from various cell differentiation

systems, comprising both single-cell RT-qPCR (scRT-qPCR) 8,10,12,35–37 and single-cell RNA-sequencing

(scRNA-seq) 38,39. We employed a likelihood-based analysis using a recent method CALISTA (Clustering And

Lineage Inference in Single-cell Transcriptomics Analysis) 40. The analysis relied on a mechanistic model of the

stochastic gene transcriptional bursts to characterize single-cell gene expression distribution. We defined a new

concept, called transcriptional uncertainty landscape, based on the cell likelihood value from CALISTA analysis

to characterize the stochastic dynamics of the gene transcription process during cell differentiation. The stochastic

gene transcriptional model enabled identifying the specific parameters or mechanisms that explain the observed

changes in the the gene transcriptional uncertainty at the single-cell level 41. For two additional single-cell datasets,

we also evaluated the single-cell RNA-velocity using the recently published Velocyto method42. The two-state

model parameter analysis, combined with RNA-velocities, provided insights into the mechanism regulating cell

fate decisions, specifically on the role of stochastic gene transcriptions in the differentiation processes and on the

possible mechanism generating this stochasticity.

RESULTS

Single-cell Transcriptional Uncertainty Landscape using CALISTA

In this work, we used CALISTA, a likelihood-based bioinformatics toolbox designed for an end-to-end

analysis of single-cell gene expression data, to evaluate the transcriptional uncertainty of each individual cell

based on its gene expression data 40. CALISTA uses the two-state model of stochastic gene transcription bursts to

characterize the distribution of mRNA counts in individual cells 43. In the model, a gene promoter stochastically

switches between ON and OFF state, and only in the ON state can gene transcription occur. The distribution of

.CC-BY-NC-ND 4.0 International license(which was not certified by peer review) is the author/funder. It is made available under aThe copyright holder for this preprintthis version posted April 25, 2020. . https://doi.org/10.1101/2020.04.23.056069doi: bioRxiv preprint

Page 5: Universality of cell differentiation trajectories revealed by a … · 1 Universality of cell differentiation trajectories revealed by a reconstruction of transcriptional uncertainty

5

mRNA depends on four model parameters: qon (the rate of promoter activation), qoff (the rate of promoter

inactivation), qt (the rate of mRNA production when the promoter is on the ON state), and qd (the rate constant of

mRNA degradation) 27,44(see Fig. 1). For example, when qoff >>qon and qoff >>qd, keeping qt /qoff fixed, mRNA are

produced through bursts of short but intense transcription, which is a typical case observed for gene transcriptions

in single cells 45,46.

Our analysis procedure using CALISTA involves four main steps: cell clustering, lineage inference, and

calculation of single-cell transcriptional uncertainty. In the analysis, each cell is assigned a likelihood value, which

is computed from the probability of its gene expression (mRNA counts) based on the mRNA distribution given

by the two-state model above. In the single-cell clustering, we employed a greedy algorithm to find single-cell

clusters that maximizes the total likelihood value for the cells. In the lineage progression inference, we connected

cell clusters sharing similar gene expression distributions – by computing cluster distances based on cell likelihood

– to form a hierarchical network. Here, we assigned each cell to an edge in the lineage progression network that

is pointing to or emanating from its respective cluster, such that its single-cell likelihood is maximized. Finally,

we computed single-cell transcriptional uncertainty as the negative logarithm of the single-cell likelihood (NLL)

value. The full detail of CALISTA can be found in a recent publication 40.

The single-cell likelihood value reflects the joint probability of its gene expression repertoire. A cell with a

low likelihood value may indicate that the gene expression of the cell is different from its neighboring cells, i.e.

the cell is an outlier. But, more interestingly, a low likelihood value may also correspond to a cell state of high

uncertainty in the gene expression. The group of cells in such high uncertainty state have gene expressions that

are dissimilar to each other, and thus, the gene expression distributions will have high entropy. As mentioned

above, in this work we used the negative logarithm of the single-cell likelihood value as a metric of single-cell

transcriptional uncertainty. By plotting the single-cell transcriptional uncertainty over the two-dimensional

projection of the single-cell transcriptomics data – for example, using the first two principal components from

Principal Component Analysis (PCA), we constructed a transcriptional uncertainty landscape in the form of a

surface plot of the NLL values. By visualizing the single-cell transcriptional uncertainty as a surface plot, we can

study the landscape of transcriptional uncertainty during cell differentiation at single-cell resolution. On the

single-cell transcriptional uncertainty surface, an aberrant cell can be easily distinguished from a cell of high

uncertainty state, since an aberrant cell will appear isolated from its nearby cells and will be located on a region

with high NLL.

.CC-BY-NC-ND 4.0 International license(which was not certified by peer review) is the author/funder. It is made available under aThe copyright holder for this preprintthis version posted April 25, 2020. . https://doi.org/10.1101/2020.04.23.056069doi: bioRxiv preprint

Page 6: Universality of cell differentiation trajectories revealed by a … · 1 Universality of cell differentiation trajectories revealed by a reconstruction of transcriptional uncertainty

6

Transcriptional uncertainty landscape of iPSC cell differentiation to cardiomyocytes

In the following, we demonstrated an application of our procedure described above to a single-cell

transcriptional dataset from cardiomyocytes differentiation from human induced pluripotent stem cells (iPSCs) 35.

The single-cell clustering of CALISTA returned five clusters 40, in good agreement with the number of cell types

reported in the original study. CALISTA identified one bifurcation event in the lineage progression, which led to

two cell lineages 35. Fig. 2a-b gives the single-cell transcriptional uncertainty landscape showing cells exiting the

initial epiblast state that is characterized by a valley in the landscape, passing through a hill of high transcriptional

uncertainty corresponding to primitive streak (PS)-like progenitor state, before ending up at one of the low

transcriptional uncertainty terminal states corresponding to either mesodermal (desired) or endodermal

(undesired) fate. As depicted in Fig, 2c, the intermediate cell cluster (cluster 2) comprising PS-like cells have

higher cell uncertainty (lower single-cell likelihood) than the other clusters. Figures 2d and 2e give the moving-

averaged uncertainty values for pseudotemporally ordered cells using a moving window of 10% of the total cells

for both endodermal and mesodermal paths, respectively. The moving-averaged transcriptional uncertainty for

the two differentiation paths follows a rise-then-fall trajectory where the peak of uncertainty coincides with the

lineage bifurcation event.

We explored whether the rise-then-fall in uncertainty is an artefact from using the two-state model to evaluate

the cell likelihood values. To this end, we implemented a modified version of the algorithm for ordering cells by

calculating the cell likelihood values using the empirical (observed) distribution, instead of the analytical

distribution from the two-state model. As shown in Supplementary Fig. S1, the transcriptional uncertainty

landscape from the modified implementation shows a strong resemblance to the original one. We also investigated

whether the number of clusters may affect the landscape, in which using too few of clusters may artificially inflate

the uncertainty due to mixing of cells from different states. We reran CALISTA by using a higher number of

clusters (set to nine based on the eigengap heuristic 47). The hill in the uncertainty landscape is again seen around

the bifurcation event upon using a higher number of cell clusters (Supplementary Fig. S2). Finally, we used a

different algorithm to cluster cells, specifically using a Laplacian-based clustering algorithm SIMLR 48, to test

whether the shape of the transcriptional uncertainty landscape changes with the clustering algorithm. The single-

cell clusters can be interpreted as the transitional states that the differentiating cells go through. Starting with the

result of SIMLR cell clustering, we then generated the lineage progression and estimated the cell likelihood values

using CALISTA. The transcriptional uncertainty landscape from SIML cell clustering has the same shape as that

.CC-BY-NC-ND 4.0 International license(which was not certified by peer review) is the author/funder. It is made available under aThe copyright holder for this preprintthis version posted April 25, 2020. . https://doi.org/10.1101/2020.04.23.056069doi: bioRxiv preprint

Page 7: Universality of cell differentiation trajectories revealed by a … · 1 Universality of cell differentiation trajectories revealed by a reconstruction of transcriptional uncertainty

7

in Fig. 2a-b, demonstrating that the transcriptional uncertainty landscape observed above is not dependent on

using CALISTA for cell clustering (Supplementary Fig. S3).

To further elucidate the role of specific genes in shaping the transcriptional uncertainty landscape, we looked

at the transcriptional uncertainty associated with individual genes. Fig. 2f depicts the NLL distribution of each

gene for the five single-cell clusters. As expected, cell in cluster 2 have generally higher NLL than those in the

other clusters. Fig. 2f clearly illustrates that within cluster 2, some genes show higher NLL values than the others

(Supplementary Figure S4). To identify the important genes related to transcriptional uncertainty, we identified

genes with NLL values exceeding a threshold 𝛿 for at least 30% of the cells in each cluster, where 𝛿 is set to 3

standard deviation above the overall mean NLL for all cells and genes in the dataset (see Methods Eq. (2)). None

of the genes in clusters 1, 4 and 5 have NLL above the threshold. Meanwhile, 16 and 8 genes in clusters 2 and 3,

respectively, pass the above criterion for high uncertainty with 4 common genes between the two gene sets

(Supplementary Table S1). Genes with high transcriptional uncertainty in cluster 2 may have functional roles in

cell fate determination. The gene set of cluster 2 includes known genes upregulated only in the PS-like state (e.g.

EOMES, GSC, MESP1 and MIXL1), as well as markers of mesodermal and endodermal cells (e.g. BMP4,

HAND1, and SOX17) 35,40 (Supplementary Figure S5). Meanwhile, the main contributors to cell uncertainty in

cluster 3 (e.g. BMP4 and MYL4 35,40) are known transition genes between PS-like cells and the final mesoderm

fate (Supplementary Figure S6). Fig. 2g depicts the protein-protein interaction (PPI) network related to the gene

set of cluster 2 using STRING (minimum required interaction score of 0.4) 49, indicating that these genes form a

strongly interconnected hub of known transcription factors and molecules involved in the signal transduction of

embryonic development (Supplementary Table S1).

Transcriptional uncertainty landscapes of cell differentiation

We further applied the procedure above to seven additional single-cell transcriptomic datasets that were

generated using scRT-qPCR 8,10,12,36,37 and scRNA-sequencing 38,39, to assess the universality of the rise-then-fall

feature of single-cell transcriptional uncertainty landscape during cell differentiation. The first of these datasets

came from 405 cells during mouse embryonic fibroblast (MEF) reprogramming into induced neural (iN) and

myogenic (M) cells 38. Like the iPSC differentiation above, the lineage progression has a single bifurcation point.

As depicted in Figure 3a, the single-cell transcriptional uncertainty increases from the initial MEF state and

reaches a peak around the bifurcation before decreasing toward two end-point cell fates. The rise-then-fall of

transcriptional uncertainty in the MEF reprogramming is in good agreement with what we observed in the iPSCs

.CC-BY-NC-ND 4.0 International license(which was not certified by peer review) is the author/funder. It is made available under aThe copyright holder for this preprintthis version posted April 25, 2020. . https://doi.org/10.1101/2020.04.23.056069doi: bioRxiv preprint

Page 8: Universality of cell differentiation trajectories revealed by a … · 1 Universality of cell differentiation trajectories revealed by a reconstruction of transcriptional uncertainty

8

differentiation above. Higher entropy of gene expression distribution in a cell population has also been reported

in the reprogramming of iPSCs13.

Next, we analyzed datasets from cell differentiation processes without a lineage bifurcation and with

multiple lineage bifurcations. Three scRT-qPCR datasets came from differentiation systems without bifurcation,

including Richard et al. study on chicken erythrocytic differentiation of T2EC cells 8, Stumpf et al. study on

differentiation of mouse embryonic stem cells (ESC) to neural progenitor cells (NPC) 10, and Moussy et al. study

during CD34+ cell differentiation 12. The single-cell clustering and lineage progression by CALISTA produced

the expected cell differentiation trajectory (see Supplementary Figure S7-9). The single-cell transcriptional

uncertainty landscapes of these three differentiation systems, as shown in Figure 3b, exhibit a rise-then-fall profile,

creating a hill that the cells traverse through in the differentiation process. A transitory increase in single-cell gene

expression uncertainty was reported either directly or indirectly in the original publications. In Richard et al. 8 and

Stumpf et al 10, the authors adopted the Shannon entropy to quantify cell-to-cell variability (uncertainty), while

Moussy et al. 12 reported an unstable transition state with ‘hesitant cells’ flipping their morphology between

polarized and round shapes before committing to the common myeloid progenitors-like fate. Morphological

uncertainty therefore corresponded to a higher transcriptional uncertainty. Note that Moussy et al. study looked

at only the initial phase of the (hematopoietic) cell differentiation, and thus, it is likely that the differentiation

process had not completed for the cells in the dataset.

The next set of single-cell gene expression data came from differentiation systems with multi-branching

lineage, including Guo et al. study during mouse embryo development from zygote to blastocyst 36, Nestorowa et

al. 39 and Moignard et al. 37 studies on hematopoietic stem cell differentiation. Figure 3c show the single-cell

transcriptional landscape for each of the datasets. For Guo et al. study, we identified 7 cell clusters and identified

two bifurcations in the lineage. Here, we observed two hills in the transcriptional uncertainty landscape, each

coinciding with a bifurcation event in the lineage progression – one at 32-cell stage (cluster 2 to cluster 3 and 4)

and another at 64-cell stage (cluster 4 to cluster 6 and 7) (see Supplementary Figure S10). For Nestorowa et al. 39

(Supplementary Figure S11) and Moignard et al. 37 (see Methods and 40) datasets, we again observed peaks in the

transcriptional uncertainty landscape that colocalize with the bifurcation points in the lineage progression.

The use of the two-state mechanistic gene transcriptional model within CALISTA enabled us to probe into a

mechanistic explanation for the observed shape of the transcriptional uncertainty landscape. Table 1 show the

pairwise Pearson correlations between the cell-averaged NLL of each cluster with two biologically interpretable

model parameters, namely transcriptional burst size (number of transcripts generated in each burst) and burst

.CC-BY-NC-ND 4.0 International license(which was not certified by peer review) is the author/funder. It is made available under aThe copyright holder for this preprintthis version posted April 25, 2020. . https://doi.org/10.1101/2020.04.23.056069doi: bioRxiv preprint

Page 9: Universality of cell differentiation trajectories revealed by a … · 1 Universality of cell differentiation trajectories revealed by a reconstruction of transcriptional uncertainty

9

frequency (occurrence of burst per unit time) 50 (see Methods). The Pearson correlations indicate that the single-

cell gene expression uncertainty increases with higher burst size and burst frequency (p-value ≤ 0.01). Higher

transcriptional burst size and frequency are associated with a lower qoff – a lower rate of promoter turning off –

and a greater qon – higher rate of promoter turning on. One possible explanation for such change in model

parameters is a higher chromatin accessibility during the transition period of cell differentiation. This finding is

consistent with the view that stem cells increase its gene expression uncertainty or stochasticity by adopting a

more open chromatin state to enable the exploration of the gene expression space 33,50–52.

Coupling cell uncertainty with RNA velocity

In a recent paper 42, La Manno and colleagues introduced the concept of RNA velocity, which involves

computing the rate of change of mRNA from the ratio of unspliced to spliced mRNA. A positive RNA velocity

indicates an induction of gene expression, while a negative RNA velocity indicates a repression of gene

expression. La Manno et al. demonstrated that RNA velocities are able to predict the trajectory of cells undergoing

a dynamical transition, such as in circadian rhythms or cell differentiation. In the following, we explored the

relationship between RNA velocities and single-cell transcriptional uncertainty.

We evaluated the single-cell transcriptional uncertainty and RNA velocity for two single-cell gene expression

datasets that were previously analyzed in La Manno et al. 42; 42. The first dataset came from human glutamatergic

neurogenesis which has a linear (non-bifurcating) lineage progression. Figure 4 (top row) depicts the cell

clustering, single-cell transcriptional uncertainty, and RNA velocities (see also Supplementary Figure S12). The

single-cell transcriptional uncertainty landscape again has the rise-then-fall shape, as in the other cell

differentiation systems discussed above. Interestingly, the same rise-then-fall profile is also seen in the RNA

velocity. As illustrated in Figure 4, the increase and decrease of the RNA velocity preceed the transcriptional

uncertainty, and the peak of RNA velocity occurs prior to those of the transcriptional uncertainty (see

Supplementary File S1 for an animated illustration). Furthermore, a gene-wise cross-correlation analysis confirms

a positive correlation between RNA velocity and single-cell transcriptional uncertainty with a delay for individual

genes (see Supplementary Figure S13).

We also compared RNA velocity and single-cell transcriptional uncertainty for another dataset from mouse

hippocampal neurogenesis with a multi-branching lineage 53. Figure 4 (bottom row) shows that like in the

neurogenesis dataset earlier, the RNA velocity increases and then decreases during cell differentiation, and the

change in the RNA precede that of the transcriptional uncertainty (see Supplementary File S2). Also, the RNA

.CC-BY-NC-ND 4.0 International license(which was not certified by peer review) is the author/funder. It is made available under aThe copyright holder for this preprintthis version posted April 25, 2020. . https://doi.org/10.1101/2020.04.23.056069doi: bioRxiv preprint

Page 10: Universality of cell differentiation trajectories revealed by a … · 1 Universality of cell differentiation trajectories revealed by a reconstruction of transcriptional uncertainty

10

velocity peaks take place before the transcriptional uncertainty peaks. The rise-then-fall dynamic of the RNA

velocity seen in the two datasets above is consistent with the view that cells engage in an exploratory stochastic

dynamics as they leave the progenitor state, and disengage this explorative mode as they reach toward the final

cell state.

DISCUSSION

Although Waddington’s epigenetic landscape was originally proposed only as a metaphor, the landscape has

helped stem cell researchers to conceptualize the cell differentiation processes through canalization of cell

lineages. As mentioned earlier, much of the existing literature on the analytical reconstruction of the epigenetic

landscape relied on either a dynamical system theory applied to a simple gene network, or a thermodynamic

interpretation based on the potential energy of a reaction 21–23. In our study, we did not make any prior assumptions

on the gene regulatory network driving the differentiation process nor on the characteristics of the landscape, such

as the existence of a stable valley or that of an energetic barrier (hill). Here, we assumed that the gene transcription

at the single-cell level occurs as stochastic transcriptional bursts, in which mRNA counts follow the equilibrium

distribution that is given by the two-state gene transcription model 43. Therefore, the transcriptional uncertainty

landscape in our study is a reflection of the dynamic evolution of gene transcriptional stochasticity within

individual cells during the cell differentiation process.

The reconstruction of the transcriptional uncertainty landscapes from 10 single-cell transcriptomic datasets

of various cell differentiation processes in our study reveals a universal rise-then-fall trajectory in which cells start

from a high potency state with a uniform gene expression pattern in the cell population, then progress through

transitional cell state(s) marked by increased transcriptional uncertainty (i.e., higher cell-to-cell variability), and

eventually reach one of possibly several final cell states with again a uniform gene expression pattern among the

cells. Furthermore, the peaks of the transcriptional uncertainty landscape colocalize with forks in the cell lineage.

The rise-then-fall in cell uncertainty agrees well with other reports from different cell differentiation systems 8–

12,54,55, suggesting that stem cells go through a transition state of high gene expression uncertainty before

committing to a particular cell fate. The existence of a hill or barrier during the intermediate stage of cell

differentiation has also been proposed in previous studies 14,31,56. In particular Moris and colleagues compared this

transition state to the activation energy barrier in chemical reactions 56. We noted however, that a hill in our

transcriptional uncertainty landscape is a reflection of a peak in the cell-to-cell gene expression variability, and

thus does not represent a resistance or barrier that a cell has to overcome.

.CC-BY-NC-ND 4.0 International license(which was not certified by peer review) is the author/funder. It is made available under aThe copyright holder for this preprintthis version posted April 25, 2020. . https://doi.org/10.1101/2020.04.23.056069doi: bioRxiv preprint

Page 11: Universality of cell differentiation trajectories revealed by a … · 1 Universality of cell differentiation trajectories revealed by a reconstruction of transcriptional uncertainty

11

In the analysis of iPSCs differentiation into cardiomyocytes 35, the key genes regulating cardiomyocyte

differentiation are among the largest contributors to the overall transcriptional uncertainty at or around the peak

in the landscape, supporting the idea that dynamic cell-to-cell variability has a functional role in cell-fate decision

making processes 21,57,58. Such an idea would be in congruence with the recent demonstration that, in a

physiologically relevant cellular system, gene expression variability is functionally linked to differentiation 57,58.

The rise-then-fall trajectory in the transcriptional uncertainty landscape are more pronounced in some datatsets

than in others. For example, in Nestorowa 39 and Moignard 37 datasets (see Figure 3c), peaks in the transcriptional

uncertainty landscape are less noticeable than in the other differentiation systems. We noted that cells in the

Nestorowa 39 and Moignard 37 studies were pre-sorted by using flow cytometry based on the expression of surface

protein markers. We posited that at least some cells in the transition state(s) might have been lost during the cell

pre-sorting since such cells might not express the chosen surface markers strongly.

Further, the correlation analysis between the cell transcriptional uncertainty and biologically meaningful rates

of the stochastic gene transcription model showed strong positive correlations with transcriptional burst size and

frequency. In agreement with the result of the correlation analysis, several studies have reported an increase in

gene transcriptional bursts during transition states in cell differentiation and other recent studies have suggested

that both burst frequency and burst size regulate gene expression levels 33,51,52. Importantly, our comparison of the

single-cell transcriptional uncertainty and the single-cell RNA velocity revealed that an increase (decrease) in

RNA velocity predicts an increase (decrease) in transcriptional uncertainty after a short delay, and that a peak of

RNA velocity preceeds that of the transcriptional uncertainty.

The aforementioned observations, while correlative in nature, points to possible biological mechanisms

underlying the universal dynamic feature of single-cell transcriptional uncertainty during cell differentiation. At

the start of the differentiation process, cells engage an exploratory search dynamics in the gene expression space

by increasing stochastic transcriptional burst size and burst frequency. The putative objective of such a stochastic

search is to optimize the cell’s gene expression pattern given its new environment. The engagement of this

stochastic exploratory mode is supported by the observed increased in the overall RNA velocity and its expected-

but-delayed effect in elevating the cell-to-cell gene expression variability (i.e. higher transcriptional uncertainty).

Increased transcriptional burst size and frequency suggest a promoter state of stochastic gene transcription that

favors the ON state (higher qon and lower qoff ).

A possible mechanism behind this exploratory search dynamics is an increase in chromatin mobility, driven by

metabolic alterations in early differentiation 59. Multiple studies have demonstrated that a mismatch between the

.CC-BY-NC-ND 4.0 International license(which was not certified by peer review) is the author/funder. It is made available under aThe copyright holder for this preprintthis version posted April 25, 2020. . https://doi.org/10.1101/2020.04.23.056069doi: bioRxiv preprint

Page 12: Universality of cell differentiation trajectories revealed by a … · 1 Universality of cell differentiation trajectories revealed by a reconstruction of transcriptional uncertainty

12

intracellular state of stem cells and their immediate environment can lead to metabolic reorganization 60–62.

More specifically, a change in the balance between glycolysis and OXPHOS metabolism has been associated to

numerous differentiation processes (see Richard et al. 63 and references therein).

Furthermore, changes in the metabolic flux state in early differentiation can modulate the activity of chromatin

modifying enzymes through their metabolic co-factors 64, or in more direct fashion 65 and alter the cell

differentiation outcome. A more dynamic state of the chromatin is associated with more variable gene expressions

due to the changes in the opening-closing dynamics (breathing) of the chromatin 66. As the cells approach the final

state, cells disengage the exploratory search mode, as the cells approach an optimal gene expression and metabolic

state associated with a chosen cell type.

The findings of our analysis fit within the paradigm of a stochastic stem cell differentiation process as

proposed in the introduction. The disordered gene expression pattern during the transition period can be seen as

an exploratory dynamics to find the optimal pattern(s) 14,17. The transcriptional uncertainty in our analysis can be

interpreted as the width of the valley in Waddington’s epigenetic landscape. If one considers the epigenetic

landscape as a depiction of the accessible gene expression subspace through with stochastic single-cell trajectories

pass during differentiation, a wider valley indicates a more variable gene expression pattern. While in the original

Waddington’s epigenetic landscape the valley naturally widens around the branching point in the cell lineage, our

analysis shows that a widening of the valley (an increase in transcriptional uncertainty) also occurs in non-

branching lineage. In other words, the increase in transcriptional uncertainty appears to be a universal feature of

the cell differentiation process, one that arises from the engagement of exploratory mode through increased

stochasticity in transcriptional bursts, as explained above.

METHODS

Main steps of CALISTA workflow

Herein, we briefly describe the main steps involved in the calculation of single-cell transcriptional uncertainty

using CALISTA (for further details see 40).

Pre-processing

Given an 𝑁 × 𝐺 single-cell expression matrix 𝑀, where N denotes the number of cells and G the number of genes,

the pre-processing in CALISTA involves two steps: a normalization of the expression data 𝑚!,# – i.e. the number

of transcripts of gene g in the n-th cell, and a selection of the most variable genes 40.

.CC-BY-NC-ND 4.0 International license(which was not certified by peer review) is the author/funder. It is made available under aThe copyright holder for this preprintthis version posted April 25, 2020. . https://doi.org/10.1101/2020.04.23.056069doi: bioRxiv preprint

Page 13: Universality of cell differentiation trajectories revealed by a … · 1 Universality of cell differentiation trajectories revealed by a reconstruction of transcriptional uncertainty

13

Cell clustering

CALISTA clustering follows a two-step procedure. The first step involves a greedy optimization strategy to find

cell clustering that maximizes the total cell likelihood, i.e. the sum of the likelihood value for all cells. The single-

cell likelihood value is computed as the joint probability of the cell’s gene expression data, which is set equal to

the product of the probabilities of the mRNA counts for the selected genes based on the mRNA distribution from

the two-state stochastic gene transcription model. To avoid issues with numerical overflow, we use the logarithm

of the cell likelihood. By performing the greedy optimization multiple times, a consensus matrix containing the

number of times two cells in the dataset are put in the same cluster, is generated.. In the second and final step,

CALISTA generates the cell cluster assignments by using k-medoids clustering based on the consensus matrix.

The final outcome of CALISTA’s clustering is the assignment of cells into K clusters and the optimal model

parameters for the two-state gene transcription model: 𝜃∗(𝑔, 𝑘) = .𝜃%!∗ , 𝜃%&&∗ , 𝜃'∗/#(

for each gene g in cluster k (for

simplicity, we set 𝜃)∗ = 1 and scale the other parameters by the degradation rate) 40.

Lineage progression inference

In CALISTA, cell lineage progression is inferred based on cluster distances – a measure of dissimilarity between

two clusters. The cluster distance of two cell clusters is defined as the average decrease in the cell likelihood value

if the cells from these two clusters are grouped as one cluster, as opposed to the original clustering. The lineage

progression graph is built by adding transition edges between pairs of clusters in increasing magnitude of cluster

distance until all clusters are connected to at least one other cluster, or based on user-specified criteria.

Single-cell transcriptional uncertainty

The last step in our analysis is to compute the final single-cell likelihood. Briefly, for each cell, we consider all

edges in the lineage progression graph that are adjacent to the cell’s respective cluster, i.e. edges that eminate

from or pointing to the cluster to which the cell belongs. The likelihood of a cell along an edge is evaluated by

interpolating the likelihood values of the cell’s gene expression using the mRNA distributions from the two

adjacent clusters. Each cell is then assigned to the edge along which its interpolated likelihood value is maximum,

and the final cell likelihood is set to this maximum value. As mentioned above, the single-cell transcriptional

uncertainty is evaluated as the negative logarithm of the cell likelihood value (NLL).

.CC-BY-NC-ND 4.0 International license(which was not certified by peer review) is the author/funder. It is made available under aThe copyright holder for this preprintthis version posted April 25, 2020. . https://doi.org/10.1101/2020.04.23.056069doi: bioRxiv preprint

Page 14: Universality of cell differentiation trajectories revealed by a … · 1 Universality of cell differentiation trajectories revealed by a reconstruction of transcriptional uncertainty

14

Pseudotimes calculation

For we can evaluate the pseudotimes for the cells according to the following procedure. First, a pseudotime is

given to each cluster with a value between 0 (initial cell state) and 1 (final cell fate). Subsequently, we determine

the linear fractional position of each cell along its respective edge at which its interpolated likelihood value is

maximum (see Single-cell transcriptional uncertainty). The pseudotime of a cell is computed by a linear

interpolation of the pseudotimes of the two clusters adjacent to its assigned edge according to the cell’s linear

fractional position on this edge.

Epigenetic landscape reconstruction

To visualize the 3D transcriptional uncertainty landscape, we apply dimensional reduction techniques such as

Principal Component Analysis (PCA) or t-SNE on the z-scored expression data, to project the gene expression of

each individual cell on two dimensional axis, which gives the x-y axis of the landscape plot. For the z axis, we

plot the NLL values. The transcriptional uncertainty landscape surface is reconstructed by estimating local

approximation of individual cell 3D coordinates on a regular 30x30 grid by using a publicly available Matlab

surface fitting code called GRIDFIT 67.

Pre-processing and analysis of single-cell expression datasets

Bargaje et al. scRT-qPCR dataset.

The dataset includes the expression profiles of 96 genes from 1896 single cells at 8 different time points (day 0,

1, 1.5, 2, 2.5, 3, 4, 5) during the differentiation of human pluripotent stem cells (iPSCs) into either mesodermal

(M) or endodermal (En) fate 35. By employing CALISTA, we obtained five cell clusters and detected a bifurcation

event, which gives rise to the two final cell fates. After lineage inference, we pseudotemporally ordered cells

along the inferred differentiation paths (for more details, see 40).

Treutlein et al. scRNA-sequencing dataset.

The dataset includes the gene expression profiles of 405 cells during reprogramming of mouse embryonic

fibroblast (MEF) into a desired induced neural (iN) and an alternative myogenic (M) cell fates 38. We pre-

processed the data using CALISTA to select the 40 most variable genes (10% of the number of cells) for the

transcriptional uncertainty analysis. CALISTA identified four different subpopulations and successfully

recovered the bifurcation event (for more details, see 40)

.CC-BY-NC-ND 4.0 International license(which was not certified by peer review) is the author/funder. It is made available under aThe copyright holder for this preprintthis version posted April 25, 2020. . https://doi.org/10.1101/2020.04.23.056069doi: bioRxiv preprint

Page 15: Universality of cell differentiation trajectories revealed by a … · 1 Universality of cell differentiation trajectories revealed by a reconstruction of transcriptional uncertainty

15

Richard et al. scRT-qPCR dataset.

The dataset contains the expression profile of 91 genes measured from 389 cells at 6 distinct time points (0, 8, 24,

33, 48, 72 h) during the differentiation of primary chicken erythrocytic progenitor cells (T2EC) 8. Following

CALISTA pre-processing step, we removed cells in which less than 75% of the genes are expressed. Then, we

selected the subset of genes with at least one non-zero expression values. A total of 354 cells and 88 genes were

considered in the transcriptional uncertainty analysis. Based on eigengap heuristics 40,47, we grouped cells into 6

optimal clusters and ordered cells along the inferred linear trajectory (see Supplementary Figure S7).

Stumpf et al. scRT-qPCR dataset.

The dataset comprises the single-cell expression of 97 genes at 7 time points (0, 24, 48, 72, 96, 120, 168 h) during

neural differentiation of mouse embryonic stem cells (E14 cell line) 10. In the data pre-processing, we excluded

cells in which less than 70% of genes are expressed. Then, we selected genes with at least one non-zero expression

values. A total of 276 cells and 93 genes were considered for for the transcriptional uncertainty analysis. Based

on eigengap heuristics 40, we grouped cells into five optimal clusters and ordered cells along the inferred linear

trajectory (Supplementary Figure S8).

Moussy et al. scRT-qPCR dataset.

The single-cell expression dataset includes normalized Ct values for 91 genes in 435 cells captured at 5 distinct

time points (0, 24, 48, 72, 96 h) during human cord blood-derived CD34+ differentiation 12. We employed

CALISTA to group cells into 7 clusters, reconstruct the developmental trajectory and calculate pseudotimes

(Supplementary Figure S9).

Guo et al. scRT-qPCR dataset.

The dataset comprises the single-cell expression values of 48 genes from 387 individual cells isolated at 4 distinct

developmental cell stages, from 8-cell stage mouse embryos to 64-blastocyst 36. By applying CALISTA, we

identified seven different subpopulations along the differentiation process, and the inferred lineage hierarchy

pinpointed two bifurcations events at 32- and 64-cell stage (Supplementary Figure S10). The timing of the lineage

bifurcations coincides with two well-known branching points: one at 32-cell stage when totipotent cells

differentiate into trophectoderm (TE) and inner cell mass (ICM), and another at 64-cell stage when ICM cells

differentiate into primitive endoderm (PE) and epiblast (E).

.CC-BY-NC-ND 4.0 International license(which was not certified by peer review) is the author/funder. It is made available under aThe copyright holder for this preprintthis version posted April 25, 2020. . https://doi.org/10.1101/2020.04.23.056069doi: bioRxiv preprint

Page 16: Universality of cell differentiation trajectories revealed by a … · 1 Universality of cell differentiation trajectories revealed by a reconstruction of transcriptional uncertainty

16

Nestorowa et al. scRNA-sequencing dataset.

The dataset comprises single-cell gene expression of 1656 cells from mouse hematopoietic stem cell

differentiation 39. We pre-processed the data by removing genes with non-zero values in less than 10% of the

cells. Then, we selected 433 most variable genes, which is 10% of the number of genes after the previous pre-

processing step, for the transcriptional uncertainty analysis 40. We set the optimal number of clusters based on the

original study 39, which reported six different subpopulations and two bifurcation events: the first one producing

common myeloid progenitor (CMP) from lymphoid-primed multipotent progenitors (LMPP), and the second one

generating granulocyte–monocyte progenitors (GMP) from megakaryocyte-erythroid progenitors (MEP)

(Supplementary Figure S11).

Moignard et al. scRT-qPCR dataset.

The dataset contains the single-cell expression level of 18 transcription factors measured in a total of 597 mouse

bone marrow cells during hematopoietic differentiation. By applying CALISTA, we successfully identified the

five subpopulations and the two branching points detected in the original study 37: long-term hematopoietic stem

cells (HSC) differentiating into megakaryocyte–erythroid progenitors (PreM) or lymphoid-primed multipotent

progenitors (LMPP); LMPP cells differentiating into granulocyte–monocyte progenitors (GMP) and common

lymphoid progenitors (CLP) (for details see 40).

Pairwise correlation analysis of transcriptional uncertainty and transcriptional burst size

and frequency.

We defined gene transcriptional burst size and burst frequency using the two-state model parameters, as follows:

𝐵𝑢𝑟𝑠𝑡𝑠𝑖𝑧𝑒 = 𝜃'𝜃%&&

(1)

𝐵𝑢𝑟𝑠𝑡𝐹𝑟𝑒𝑞𝑢𝑒𝑛𝑐𝑦 = 𝜃%! (2)

The burst sizes and burst frequencies are evaluated using the parameters 𝜃∗(𝑔, 𝑘) = .𝜃%!∗ , 𝜃%&&∗ , 𝜃'∗/#(

obtained from

single-cell clustering analysis of CALISTA. Meanwhile, the average gene-wise NLL values for each single-cell

cluster was computed as:

where 𝑁𝐿𝐿#! is the negative log-likelihood of cell n based only on the expression of gene g, and Nk is the total

number of cells in cluster k.

𝑁𝐿𝐿#( =∑ +,,!"#$"%&+$

(3)

.CC-BY-NC-ND 4.0 International license(which was not certified by peer review) is the author/funder. It is made available under aThe copyright holder for this preprintthis version posted April 25, 2020. . https://doi.org/10.1101/2020.04.23.056069doi: bioRxiv preprint

Page 17: Universality of cell differentiation trajectories revealed by a … · 1 Universality of cell differentiation trajectories revealed by a reconstruction of transcriptional uncertainty

17

RNA VELOCITY ANALYSIS

Cells and genes were first filtered based on the pre-processing strategy in the original publication by La Manno

and colleagues 42, which resulted in a total of 1720 cells and 1448 genes from human glutamatergic neurogenesis,

and a total of 18140 cells and 2141 genes from mouse hippocampus dataset. We further reduced the number of

genes to only the top 500 highly variable genes for the transcriptional uncertainty analysis. The cell cluster

assignments generated by Velocyto – the algorithm for computing RNA velocity from the original publication –

were considered, instead of using CALISTA. Based on the clustering, we employed CALISTA to generate the

lineage progression and cell pseudotimes (Supplementary Figure S12). The RNA velocity and transcriptional

uncertainty values for the top 500 genes were calculated by employing Velocyto and CALISTA, respectively.

The cell-wise RNA velocity was set to the Euclidean norm of the vector of RNA velocities for each cell, while

the cell-wise NLLs was computed according to:

CODE AVAILABILITY

A MATLAB and R version of CALISTA used in this study is freely available from the following website:

https://www.cabselab.com/calista. All additional data are available from the corresponding author upon request.

DATA AVAILABILITY

All the public single cell data sets analysed in this study are available from the original publications.

COMPETING INTERESTS

The authors declare that they have no competing interests.

FUNDING

This work was supported by the Swiss National Science Foundation (grant number 157154 and 176279) and ANR

research grant SinCity (grant number ANR-17-CE12-0031-01).

𝑁𝐿𝐿!( =∑ +,,!"'((!%&

-.. (4)

.CC-BY-NC-ND 4.0 International license(which was not certified by peer review) is the author/funder. It is made available under aThe copyright holder for this preprintthis version posted April 25, 2020. . https://doi.org/10.1101/2020.04.23.056069doi: bioRxiv preprint

Page 18: Universality of cell differentiation trajectories revealed by a … · 1 Universality of cell differentiation trajectories revealed by a reconstruction of transcriptional uncertainty

18

ACKNOWLEDGEMENTS

We would like to thank all members of the SBDM team for lively discussions. We also thank the BioSyL

Federation and the LabEx Ecofect (ANR-11-LABX-0048) of the University of Lyon for inspiring scientific

events.

.CC-BY-NC-ND 4.0 International license(which was not certified by peer review) is the author/funder. It is made available under aThe copyright holder for this preprintthis version posted April 25, 2020. . https://doi.org/10.1101/2020.04.23.056069doi: bioRxiv preprint

Page 19: Universality of cell differentiation trajectories revealed by a … · 1 Universality of cell differentiation trajectories revealed by a reconstruction of transcriptional uncertainty

19

REFERENCES

1. Ohno, S., Nagai, Y., Ciccarese, S. & Iwata, H. Testis-Organizing H-Y Antigen and the Primary Sex-

Determining Mechanism of Mammals. Proc. 1978 Laurentian Horm. Conf. 449–476 (1979)

doi:10.1016/B978-0-12-571135-7.50014-X.

2. Herskowitz, I. A regulatory hierarchy for cell specialization in yeast. Nature 342, 749–757 (1989).

3. Lewis, E. B. Clusters of Master Control Genes Regulate the Development of Higher Organisms. JAMA J.

Am. Med. Assoc. 267, 1524 (1992).

4. Davis, R. L., Weintraub, H. & Lassar, A. B. Expression of a single transfected cDNA converts fibroblasts

to myoblasts. Cell 51, 987–1000 (1987).

5. Whyte, W. A. et al. Master transcription factors and mediator establish super-enhancers at key cell identity

genes. Cell 153, 307–19 (2013).

6. Papili Gao, N., Ud-Dean, S. M. M., Gandrillon, O. & Gunawan, R. SINCERITIES: Inferring gene

regulatory networks from time-stamped single cell transcriptional expression profiles. Bioinformatics

(2017) doi:10.1093/bioinformatics/btx575.

7. Sartorelli, V. & Puri, P. L. Shaping Gene Expression by Landscaping Chromatin Architecture: Lessons

from a Master. Mol. Cell 71, 375–388 (2018).

8. Richard, A. et al. Single-Cell-Based Analysis Highlights a Surge in Cell-to-Cell Molecular Variability

Preceding Irreversible Commitment in a Differentiation Process. PLOS Biol. 14, e1002585 (2016).

9. Mojtahedi, M. et al. Cell Fate Decision as High-Dimensional Critical State Transition. PLOS Biol. 14,

e2000640 (2016).

10. Stumpf, P. S. et al. Stem Cell Differentiation as a Non-Markov Stochastic Process. Cell Syst. 5, 268-

282.e7 (2017).

11. Semrau, S. et al. Dynamics of lineage commitment revealed by single-cell transcriptomics of

differentiating embryonic stem cells. Nat. Commun. 8, 1096 (2017).

12. Moussy, A. et al. Integrated time-lapse and single-cell transcription studies highlight the variable and

dynamic nature of human hematopoietic cell fate commitment. PLOS Biol. 15, e2001867 (2017).

13. Buganim, Y. et al. Single-Cell Expression Analyses during Cellular Reprogramming Reveal an Early

Stochastic and a Late Hierarchic Phase. Cell 150, 1209–1222 (2012).

14. Braun, E. The unforeseen challenge: from genotype-to-phenotype in cell populations. Reports Prog. Phys.

78, 036602 (2015).

.CC-BY-NC-ND 4.0 International license(which was not certified by peer review) is the author/funder. It is made available under aThe copyright holder for this preprintthis version posted April 25, 2020. . https://doi.org/10.1101/2020.04.23.056069doi: bioRxiv preprint

Page 20: Universality of cell differentiation trajectories revealed by a … · 1 Universality of cell differentiation trajectories revealed by a reconstruction of transcriptional uncertainty

20

15. Kupiec, J. J. A chance-selection model for cell differentiation. Cell Death Differ. 3, 385–90 (1996).

16. Kupiec, J. J. A Darwinian theory for the origin of cellular differentiation. Mol. Gen. Genet. MGG 255,

201–208 (1997).

17. Paldi, A. Stochastic gene expression during cell differentiation: order from disorder? Cell. Mol. Life Sci.

60, 1775–1778 (2003).

18. Gupta, P. B. et al. Stochastic state transitions give rise to phenotypic equilibrium in populations of cancer

cells. Cell 146, 633–44 (2011).

19. Thomas, P., Popović, N. & Grima, R. Phenotypic switching in gene regulatory networks. Proc. Natl.

Acad. Sci. U. S. A. 111, 6994–9 (2014).

20. Huang, S. Non-genetic heterogeneity of cells in development: more than just noise. Development 136,

3853–3862 (2009).

21. Rebhahn, J. A. et al. An animated landscape representation of CD4+ T-cell differentiation, variability,

and plasticity: Insights into the behavior of populations versus cells. Eur. J. Immunol. 44, 2216–2229

(2014).

22. Bhattacharya, S., Zhang, Q. & Andersen, M. E. A deterministic map of Waddington’s epigenetic

landscape for cell fate specification. BMC Syst. Biol. 5, 85 (2011).

23. Brackston, R. D., Lakatos, E. & Stumpf, M. P. H. Transition state characteristics during cell

differentiation. PLOS Comput. Biol. 14, e1006405 (2018).

24. Ud-Dean, S. M. M. & Gunawan, R. Ensemble inference and inferability of gene regulatory networks.

PLoS One 9, e103812 (2014).

25. Bonnaffoux, A. et al. WASABI: A dynamic iterative framework for gene regulatory network inference.

BMC Bioinformatics 20, 220 (2019).

26. Ud-Dean, S. M. M. & Gunawan, R. Optimal design of gene knockout experiments for gene regulatory

network inference. Bioinformatics 32, 875–883 (2016).

27. Herbach, U., Bonnaffoux, A., Espinasse, T. & Gandrillon, O. Inferring gene regulatory networks from

single-cell data: a mechanistic approach. BMC Syst. Biol. 11, 105 (2017).

28. Qiu, X. et al. Inferring Causal Gene Regulatory Networks from Coupled Single-Cell Expression

Dynamics Using Scribe. Cell Syst. 10, 265-274.e11 (2020).

29. Lv, C., Li, X., Li, F. & Li, T. Constructing the energy landscape for genetic switching system driven by

intrinsic noise. PLoS One 9, e88167 (2014).

.CC-BY-NC-ND 4.0 International license(which was not certified by peer review) is the author/funder. It is made available under aThe copyright holder for this preprintthis version posted April 25, 2020. . https://doi.org/10.1101/2020.04.23.056069doi: bioRxiv preprint

Page 21: Universality of cell differentiation trajectories revealed by a … · 1 Universality of cell differentiation trajectories revealed by a reconstruction of transcriptional uncertainty

21

30. Guo, J. & Zheng, J. HopLand: single-cell pseudotime recovery using continuous Hopfield network-based

modeling of Waddington’s epigenetic landscape. Bioinformatics 33, i102–i109 (2017).

31. Fard, A. T., Srihari, S., Mar, J. C. & Ragan, M. A. Not just a colourful metaphor: modelling the landscape

of cellular development using Hopfield networks. npj Syst. Biol. Appl. 2, 16001 (2016).

32. Zwiessele, M. & Lawrence, N. D. Topslam: Waddington Landscape Recovery for Single Cell

Experiments. bioRxiv 057778 (2017) doi:10.1101/057778.

33. Zhang, J. & Zhou, T. Topographer Reveals Stochastic Dynamics of Cell Fate Decisions from Single-Cell

RNA-Seq Data. bioRxiv 251207 (2018) doi:10.1101/251207.

34. Shi, J., Teschendorff, A. E., Chen, W., Chen, L. & Li, T. Quantifying Waddington’s epigenetic landscape:

a comparison of single-cell potency measures. Brief. Bioinform. (2018) doi:10.1093/bib/bby093.

35. Bargaje, R. et al. Cell population structure prior to bifurcation predicts efficiency of directed

differentiation in human induced pluripotent cells. Proc. Natl. Acad. Sci. U. S. A. 114, 2271–2276 (2017).

36. Guo, G. et al. Resolution of cell fate decisions revealed by single-cell gene expression analysis from

zygote to blastocyst. Dev. Cell 18, 675–85 (2010).

37. Moignard, V. et al. Characterization of transcriptional networks in blood stem and progenitor cells using

high-throughput single-cell gene expression analysis. Nat. Cell Biol. 15, 363–72 (2013).

38. Treutlein, B. et al. Dissecting direct reprogramming from fibroblast to neuron using single-cell RNA-seq.

Nature 534, 391–395 (2016).

39. Nestorowa, S. et al. A single-cell resolution map of mouse hematopoietic stem and progenitor cell

differentiation. Blood 128, e20-31 (2016).

40. Papili Gao, N., Hartmann, T., Fang, T. & Gunawan, R. CALISTA: Clustering and LINEAGE Inference

in Single-Cell Transcriptional Analysis. Front. Bioeng. Biotechnol. 8, 18 (2020).

41. Coulon, A., Gandrillon, O. & Beslon, G. On the spontaneous stochastic dynamics of a single gene:

complexity of the molecular interplay at the promoter. BMC Syst. Biol. 4, 2 (2010).

42. La Manno, G. et al. RNA velocity of single cells. Nature 560, 494–498 (2018).

43. Peccoud, J. & Ycart, B. Markovian Modeling of Gene-Product Synthesis. Theor. Popul. Biol. 48, 222–

234 (1995).

44. Kim, J. & Marioni, J. C. Inferring the kinetics of stochastic gene expression from single-cell RNA-

sequencing data. Genome Biol. 14, R7 (2013).

45. Munsky, B., Neuert, G. & Van Oudenaarden, A. Using gene expression noise to understand gene

.CC-BY-NC-ND 4.0 International license(which was not certified by peer review) is the author/funder. It is made available under aThe copyright holder for this preprintthis version posted April 25, 2020. . https://doi.org/10.1101/2020.04.23.056069doi: bioRxiv preprint

Page 22: Universality of cell differentiation trajectories revealed by a … · 1 Universality of cell differentiation trajectories revealed by a reconstruction of transcriptional uncertainty

22

regulation. Science vol. 336 183–187 (2012).

46. Suter, D. M. et al. Mammalian Genes Are Transcribed with Widely Different Bursting Kinetics. Science

(80-. ). 332, 472–474 (2011).

47. von Luxburg, U. A tutorial on spectral clustering. Stat. Comput. 17, 395–416 (2007).

48. Wang, B., Zhu, J., Pierson, E., Ramazzotti, D. & Batzoglou, S. Visualization and analysis of single-cell

RNA-seq data by kernel-based similarity learning. Nat. Publ. Gr. 14, (2017).

49. Szklarczyk, D. et al. STRING v10: protein-protein interaction networks, integrated over the tree of life.

Nucleic Acids Res. 43, D447-52 (2015).

50. Nicolas, D., Phillips, N. E. & Naef, F. What shapes eukaryotic transcriptional bursting? Mol. Biosyst. 13,

1280–1290 (2017).

51. Antolović, V., Miermont, A., Corrigan, A. M. & Chubb, J. R. Generation of Single-Cell Transcript

Variability by Repression. Curr. Biol. 27, 1811-1817.e3 (2017).

52. Fritzsch, C. et al. Estrogen-dependent control and cell-to-cell variability of transcriptional bursting. Mol

Syst Biol 14, 7678 (2018).

53. Kriegstein, A. & Alvarez-Buylla, A. The Glial Nature of Embryonic and Adult Neural Stem Cells. (2009)

doi:10.1146/annurev.neuro.051508.135600.

54. Han, X. et al. Construction of a human cell landscape at single-cell level. Nature (2020)

doi:10.1038/s41586-020-2157-4.

55. Ye, Y., Yang, Z. & Lei, J. Using single-cell entropy to describe the dynamics of reprogramming and

differentiation of induced pluripotent stem cells. bioRxiv 2020.04.13.040311 (2020)

doi:10.1101/2020.04.13.040311.

56. Moris, N., Pina, C. & Arias, A. M. Transition states and cell fate decisions in epigenetic landscapes. Nat.

Rev. Genet. 17, 693–703 (2016).

57. Moris, N. et al. Histone Acetyltransferase KAT2A Stabilizes Pluripotency with Control of Transcriptional

Heterogeneity. Stem Cells 36, 1828–1838 (2018).

58. Guillemin, A., Duchesne, R., Crauste, F., Gonin-Giraud, S. & Gandrillon, O. Drugs modulating stochastic

gene expression affect the erythroid differentiation process. PLoS One 14, e0225166 (2019).

59. Paldi, A. What makes the cell differentiate? Prog. Biophys. Mol. Biol. 110, 41–43 (2012).

60. Argüello-Miranda, O., Liu, Y., Wood, N. E., Kositangool, P. & Doncic, A. Integration of Multiple

Metabolic Signals Determines Cell Fate Prior to Commitment. Mol. Cell 71, 733-744.e11 (2018).

.CC-BY-NC-ND 4.0 International license(which was not certified by peer review) is the author/funder. It is made available under aThe copyright holder for this preprintthis version posted April 25, 2020. . https://doi.org/10.1101/2020.04.23.056069doi: bioRxiv preprint

Page 23: Universality of cell differentiation trajectories revealed by a … · 1 Universality of cell differentiation trajectories revealed by a reconstruction of transcriptional uncertainty

23

61. Gu, W. et al. Glycolytic Metabolism Plays a Functional Role in Regulating Human Pluripotent Stem Cell

State. Cell Stem Cell 19, 476–490 (2016).

62. Folmes, C. D. L. et al. Somatic oxidative bioenergetics transitions into pluripotency-dependent glycolysis

to facilitate nuclear reprogramming. Cell Metab. 14, 264–71 (2011).

63. Richard, A. et al. Erythroid differentiation displays a peak of energy consumption concomitant with

glycolytic metabolism rearrangements. PLoS One 14, e0221472 (2019).

64. Moussaieff, A. et al. Glycolysis-Mediated Changes in Acetyl-CoA and Histone Acetylation Control the

Early Differentiation of Embryonic Stem Cells. Cell Metab. 21, 392–402 (2015).

65. Zhang, D. et al. Metabolic regulation of gene expression by histone lactylation. Nature 574, 575–580

(2019).

66. Zwaka, T. P. Breathing chromatin in pluripotent stem cells. Developmental Cell vol. 10 1–2 (2006).

67. John D’Errico. Surface Fitting using gridfit. MATLAB Central File Exchange

https://www.mathworks.com/matlabcentral/fileexchange/8998-surface-fitting-using-gridfit (2020).

.CC-BY-NC-ND 4.0 International license(which was not certified by peer review) is the author/funder. It is made available under aThe copyright holder for this preprintthis version posted April 25, 2020. . https://doi.org/10.1101/2020.04.23.056069doi: bioRxiv preprint

Page 24: Universality of cell differentiation trajectories revealed by a … · 1 Universality of cell differentiation trajectories revealed by a reconstruction of transcriptional uncertainty

24

FIGURE AND TABLE LEGENDS

Figure 1. Single-cell transcriptional uncertainty landscape. The illustration depicts the landscape of single-

cell transcriptional uncertainty during a differentiation process over the (pseudo) time (from blue to yellow). Each

dot corresponds to a cell in the single-cell transcriptomic dataset. Cells start their journey from a valley in the

landscape, through a hill, before ending at one of the final valleys / states.

Initial stateTransition state

Final states

Nega

tive

log

likel

ihoo

d

OFF

mRNAx

θoff

θt

θd

θon

ON

Two-state model

.CC-BY-NC-ND 4.0 International license(which was not certified by peer review) is the author/funder. It is made available under aThe copyright holder for this preprintthis version posted April 25, 2020. . https://doi.org/10.1101/2020.04.23.056069doi: bioRxiv preprint

Page 25: Universality of cell differentiation trajectories revealed by a … · 1 Universality of cell differentiation trajectories revealed by a reconstruction of transcriptional uncertainty

25

Figure 2. Analysis of single-cell transcriptional profiles during iPSC differentiation into cardiomyocytes.

The single-cell gene expression dataset was taken from the study of Bargajeet al. 35 (a-b) Epigenetic landscape

plots estimates by CALISTA. Each dot on the landscape represents a cell where the colors indicate (a) the cell

cluster, (b) pseudotime (from dark blue to yellow). The x-y axes of the landscape plots correspond to the first and

second principal component (PC) coordinates, respectively, of the single-cell transcriptomic data. (c) Boxplots of

NLL values for each single-cell cluster. (d-e) Moving-window average NLL along (d) endoderm and (e)

mesoderm fate trajectory. (f) NLL of each gene and cell for every single-cell cluster. (g) Protein-protein interaction

network of top variable genes inferred by STRING 49. Blue nodes represent transcription factors, while red nodes

denote proteins involved in signal transduction. The width of the edges denotes the confidence for the inferred

relationship (thicker edge = higher confidence).

EM

En

PS

0 200 400 600 800 1000Cell Ordering

150

200

250

300

350

400

450

500

550

600

650

Nega

tive

log

likel

ihoo

d

Moving averaged cell-logP after cell ordering

Clusters in path: 1 2 5

0 200 400 600 800 1000 1200Cell Ordering

150

200

250

300

350

400

450

500

550

600

650

Nega

tive

log

likel

ihoo

d

Moving averaged cell-logP after cell ordering

Clusters in path: 1 2 3 4

aNL

L

NLL

NLL

NLL

Cluster Cell ordering Cell ordering

PC1 PC1

PC2PC2

NLL

c d e

b

Cells

Cluster 2 Cluster 3 Cluster 4 Cluster 5

NLL

NLL

NLL

NLL

NLL

Cluster 1

CellsCells

CellsCells

GenesGenes Genes

GenesGenes

f

g

.CC-BY-NC-ND 4.0 International license(which was not certified by peer review) is the author/funder. It is made available under aThe copyright holder for this preprintthis version posted April 25, 2020. . https://doi.org/10.1101/2020.04.23.056069doi: bioRxiv preprint

Page 26: Universality of cell differentiation trajectories revealed by a … · 1 Universality of cell differentiation trajectories revealed by a reconstruction of transcriptional uncertainty

26

Figure 3. CALISTA analysis of single-cell expression data. (a-c) Landscape plots (based on cell clusters and

pseudotime) and moving-averaged NLLs for each differentiation path of (a) single-branching trajectory (Treutlein

dataset 38), (b) linear trajectories (Richard 8, Stumpf 10, and Moussy 12 datasets), (c) multi-braching trajectories

(Guo 36, Nestorowa 39, and Moignard 37 datasets). Green and red vertical arrows in moving-averaged NLL plots

indicate the first and second peak in cell uncertainty, respectively.

0 50 100 150 200 250Cell Ordering

140

160

180

200

220

240

260

280

Nega

tive

log

likel

ihoo

d

Moving averaged cell-logP after cell ordering

Clusters in path: 1 2 3

0 50 100 150 200 250 300Cell Ordering

140

160

180

200

220

240

260

280

Nega

tive

log

likel

ihoo

d

Moving averaged cell-logP after cell ordering

Clusters in path: 1 2 4

Treu

tlein

0 50 100 150 200Cell Ordering

250

300

350

400

450

500

Nega

tive

log

likel

ihoo

d

Moving averaged cell-logP after cell ordering

Clusters in path: 1 2 3 4 5

0 50 100 150 200 250 300Cell Ordering

200

300

400

500

600

700

800

Nega

tive

log

likel

ihoo

d

Moving averaged cell-logP after cell ordering

Clusters in path: 1 2 3 4 5 6

0 100 200 300Cell Ordering

200

250

300

350

400

450

500

550

600

Nega

tive

log

likel

ihoo

d

Moving averaged cell-logP after cell ordering

Clusters in path: 2 1 4 3 5 6 7

Stum

pfRi

char

dM

ouss

y

0 50 100 150 200Cell Ordering

140

160

180

200

220

240

260

280

Nega

tive

log

likel

ihoo

d

Moving averaged cell-logP after cell ordering

Clusters in path: 1 2 3 5

0 20 40 60 80 100 120 140 160 180Cell Ordering

140

160

180

200

220

240

260

280

Nega

tive

log

likel

ihoo

d

Moving averaged cell-logP after cell ordering

Clusters in path: 1 2 4 7

0 20 40 60 80 100 120 140 160Cell Ordering

140

160

180

200

220

240

260

280

Nega

tive

log

likel

ihoo

d

Moving averaged cell-logP after cell ordering

Clusters in path: 1 2 4 6

0 20 40 60 80 100 120 140 160 180 200Cell Ordering

80

90

100

110

120

130

140

150

Nega

tive

log

likel

ihoo

d

Moving averaged cell-logP after cell ordering

Clusters in path: 1 3

0 50 100 150 200 250 300Cell Ordering

80

90

100

110

120

130

140

150

Nega

tive

log

likel

ihoo

d

Moving averaged cell-logP after cell ordering

Clusters in path: 1 2 4

0 50 100 150 200 250 300Cell Ordering

80

90

100

110

120

130

140

150

Nega

tive

log

likel

ihoo

d

Moving averaged cell-logP after cell ordering

Clusters in path: 1 2 5

0 100 200 300 400 500 600Cell Ordering

1800

1900

2000

2100

2200

2300

2400

2500

2600

Nega

tive

log

likel

ihoo

d

Moving averaged cell-logP after cell ordering

Clusters in path: 1 3

0 100 200 300 400 500 600 700 800 900Cell Ordering

1800

1900

2000

2100

2200

2300

2400

2500

2600

Nega

tive

log

likel

ihoo

d

Moving averaged cell-logP after cell ordering

Clusters in path: 1 2 4 6

0 100 200 300 400 500 600Cell Ordering

1800

1900

2000

2100

2200

2300

2400

2500

2600

Nega

tive

log

likel

ihoo

d

Moving averaged cell-logP after cell ordering

Clusters in path: 1 2 5

Guo

Nes

toro

wa

Moi

gnar

d

a

b

c

iN

MMEF

MEF -> iN

HSC MPPLMPP

CMPGMP

MEP

GMP

PreMCLP

HSC

EPE

TE

8C

MEF -> M

ESC

NPC

HSC

CMP

HSC -> CMP

ESC -> NPC

8C/16C -> TE 8C/16C -> ICM -> E

ICM

8C/16C -> ICM -> PE

HSC/MPP -> LMPP HSC/MPP ->CMP -> GMPHSC/MPP ->CMP -> MEP

HSC -> PreM HSC -> LMPP -> CLP

LMPP

HSC -> LMPP -> GMP

T2EC (0h)

T2EC (72h)

T2EC (0h) -> T2EC (72h)

16C

.CC-BY-NC-ND 4.0 International license(which was not certified by peer review) is the author/funder. It is made available under aThe copyright holder for this preprintthis version posted April 25, 2020. . https://doi.org/10.1101/2020.04.23.056069doi: bioRxiv preprint

Page 27: Universality of cell differentiation trajectories revealed by a … · 1 Universality of cell differentiation trajectories revealed by a reconstruction of transcriptional uncertainty

27

Figure 4. Comparison between RNA Velocities estimated using Velocyto 42 and CALISTA NLL values.

(Top row) Human glutamatergic neurogenesis in La Manno et al. study 42. (Bottom row) Mouse hippocampal

neurogenesis in Kreigstein et al. study 5353. (First column) Cell clustering assignments evaluated from Velocyto.

Normalized values for Euclidean norm of RNA velocities (2nd column), CALISTA single-cell transcriptional

uncertainty (NLL; 3rd column). The colors in the first column indicate the cell clusters, and those in the second-

third columns indicate the normalized cell-wise RNA velocities and NLL values respectively.

-20 -10 0 10PC1

-15

-10

-5

0

5

10

15PC

2

-20 -10 0 10PC1

-15

-10

-5

0

5

10

15

PC2

-20 -10 0 10PC1

-15

-10

-5

0

5

10

15

PC2

-20 -10 0 10 20TSNE1

-30

-20

-10

0

10

20

30

TSNE2

-20 -10 0 10 20TSNE1

-30

-20

-10

0

10

20

30

TSNE2

-20 -10 0 10 20TSNE1

-30

-20

-10

0

10

20

30

TSNE2

RNA Velocity NLLCell ClustersLa

Man

noKr

eigs

tein

Radial glia

Neuroblast

Immature neuron

Neuron

Astrocyte OPC

CA2-3-4

CA

Radial glia

nlPC

CA1-Sub

Nbl

Granule

ImmGranule

0

1

RNA

Velo

city/

NLL

a b c

.CC-BY-NC-ND 4.0 International license(which was not certified by peer review) is the author/funder. It is made available under aThe copyright holder for this preprintthis version posted April 25, 2020. . https://doi.org/10.1101/2020.04.23.056069doi: bioRxiv preprint

Page 28: Universality of cell differentiation trajectories revealed by a … · 1 Universality of cell differentiation trajectories revealed by a reconstruction of transcriptional uncertainty

28

Table 1. Pairwise correlation coefficients between transcriptional uncertainty and transcriptional burst

frequency / burst size.

Correlation with Transcriptional Uncertainty (p-value ≤ 0.01 in red boldface):

Burst Frequency Burst Size

Bargaje et al. 0.74 0.70

Treutlein et al. 0.71 0.64

Richard et al. 0.68 0.55

Stumpf et al. 0.68 0.88

Moussy et al. 0.40 0.81

Guo et al. 0.75 0.82

Nestorowa et al. 0.73 0.71

Moignard et al. 0.04 0.83

La Manno et al. 0.78 0.55

Kreigstein et al. 0.77 0.32

.CC-BY-NC-ND 4.0 International license(which was not certified by peer review) is the author/funder. It is made available under aThe copyright holder for this preprintthis version posted April 25, 2020. . https://doi.org/10.1101/2020.04.23.056069doi: bioRxiv preprint